Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Brain ; 147(5): 1856-1870, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38146224

RESUMEN

Alterations in the extracellular matrix are common in patients with epilepsy and animal models of epilepsy, yet whether they are the cause or consequence of seizures and epilepsy development is unknown. Using Theiler's murine encephalomyelitis virus (TMEV) infection-induced model of acquired epilepsy, we found de novo expression of chondroitin sulfate proteoglycans (CSPGs), a major extracellular matrix component, in dentate gyrus (DG) and amygdala exclusively in mice with acute seizures. Preventing the synthesis of CSPGs specifically in DG and amygdala by deletion of the major CSPG aggrecan reduced seizure burden. Patch-clamp recordings from dentate granule cells revealed enhanced intrinsic and synaptic excitability in seizing mice that was significantly ameliorated by aggrecan deletion. In situ experiments suggested that dentate granule cell hyperexcitability results from negatively charged CSPGs increasing stationary cations on the membrane, thereby depolarizing neurons, increasing their intrinsic and synaptic excitability. These results show increased expression of CSPGs in the DG and amygdala as one of the causal factors for TMEV-induced acute seizures. We also show identical changes in CSPGs in pilocarpine-induced epilepsy, suggesting that enhanced CSPGs in the DG and amygdala may be a common ictogenic factor and potential therapeutic target.


Asunto(s)
Amígdala del Cerebelo , Proteoglicanos Tipo Condroitín Sulfato , Giro Dentado , Convulsiones , Animales , Giro Dentado/metabolismo , Amígdala del Cerebelo/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Ratones , Convulsiones/metabolismo , Masculino , Theilovirus , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Ratones Noqueados , Agrecanos/metabolismo , Neuronas/metabolismo
2.
Nat Rev Neurosci ; 20(5): 282-297, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30792501

RESUMEN

Epilepsy is a neurological disorder afflicting ~65 million people worldwide. It is caused by aberrant synchronized firing of populations of neurons primarily due to imbalance between excitatory and inhibitory neurotransmission. Hence, the historical focus of epilepsy research has been neurocentric. However, the past two decades have enjoyed an explosion of research into the role of glia in supporting and modulating neuronal activity, providing compelling evidence of glial involvement in the pathophysiology of epilepsy. The mechanisms by which glia, particularly astrocytes and microglia, may contribute to epilepsy and consequently could be harnessed therapeutically are discussed in this Review.


Asunto(s)
Epilepsia/fisiopatología , Neuroglía/fisiología , Neuronas/fisiología , Animales , Humanos , Transmisión Sináptica/fisiología
3.
Arterioscler Thromb Vasc Biol ; 42(4): e96-e114, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35139658

RESUMEN

BACKGROUND: Vascular pericytes stabilize blood vessels and contribute to their maturation, while playing other key roles in microvascular function. Nevertheless, relatively little is known about involvement of their precursors in the earliest stages of vascular development, specifically during vasculogenesis. METHODS: We combined high-power, time-lapse imaging with transcriptional profiling of emerging pericytes and endothelial cells in reporter mouse and cell lines. We also analyzed conditional transgenic animals deficient in Cx43/Gja1 (connexin 43/gap junction alpha-1) expression within Ng2+ cells. RESULTS: A subset of Ng2-DsRed+ cells, likely pericyte/mural cell precursors, arose alongside endothelial cell differentiation and organization and physically engaged vasculogenic endothelium in vivo and in vitro. We found no overlap between this population of differentiating pericyte/mural progenitors and other lineages including hemangiogenic and neuronal/glial cell types. We also observed cell-cell coupling and identified Cx43-based gap junctions contributing to pericyte-endothelial cell precursor communication during vascular assembly. Genetic loss of Cx43/Gja1 in Ng2+ pericyte progenitors compromised embryonic blood vessel formation in a subset of animals, while surviving mutants displayed little-to-no vessel abnormalities, suggesting a resilience to Cx43/Gja1 loss in Ng2+ cells or potential compensation by additional connexin isoforms. CONCLUSIONS: Together, our data suggest that a distinct pericyte lineage emerges alongside vasculogenesis and directly communicates with the nascent endothelium via Cx43 during early vessel formation. Cx43/Gja1 loss in pericyte/mural cell progenitors can induce embryonic vessel dysmorphogenesis, but alternate connexin isoforms may be able to compensate. These data provide insight that may reshape the current framework of vascular development and may also inform tissue revascularization/vascularization strategies.


Asunto(s)
Conexina 43 , Pericitos , Animales , Diferenciación Celular , Conexina 43/genética , Conexinas/genética , Células Endoteliales , Ratones
4.
Epilepsia ; 63(4): 844-854, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35132640

RESUMEN

OBJECTIVE: Previously, we reported that inhibition of the astrocytic cystine/glutamate antiporter system xc- (SXC), using sulfasalazine (SAS), decreased evoked excitatory signaling in three distinct hyperexcitability models ex vivo. The current study expands on this work by evaluating the in vivo efficacy of SAS in decreasing astrogliosis-mediated seizure burden seen in the beta-1 integrin knockout (B1KO) model. METHODS: Video-EEG (electroencephalography) monitoring (24/7) was obtained using Biopac EEG acquisition hardware and software. EEG spectral analysis was performed using MATLAB. SAS was used at an equivalence of doses taken by Crohn's disease patients. Whole-cell patch-clamp recordings were made from cortical layer 2/3 pyramidal neurons. RESULTS: We report that 100% of B1KO mice that underwent 24/7 video-EEG monitoring developed spontaneous recurrent seizures and that intraperitoneal administration of SAS significantly reduced seizure frequency in B1KOs compared to B1KOs receiving sham saline. Spectral analysis found an acute reduction in EEG power following SAS treatment in B1KOs; however, this effect was not observed in nonepileptic control mice receiving SAS. Finally, whole-cell recordings from SXC knockout mice had hyperpolarized neurons and SXC-B1 double knockouts fired significantly less action potentials in response to current injection compared to B1KOs with SXC. SIGNIFICANCE: To devise effective strategies in finding relief for one-in-three patients with epilepsy who experience drug-resistant epilepsy we must continue to explore the mechanisms regulating glutamate homeostasis. This study explored the efficacy of targeting an astrocytic glutamate antiporter, SXC, as a novel antiepileptic drug (AED) target and further characterized a unique mouse model in which chronic astrogliosis is sufficient to induce spontaneous seizures and epilepsy. These findings may serve as a foundation to further assess the potential for SAS or inform the development of more potent and specific compounds that target SXC as a novel treatment for epilepsy.


Asunto(s)
Epilepsia , Sulfasalazina , Animales , Antiportadores , Electroencefalografía , Epilepsia/tratamiento farmacológico , Gliosis , Ácido Glutámico , Humanos , Ratones , Convulsiones/tratamiento farmacológico , Sulfasalazina/farmacología , Sulfasalazina/uso terapéutico
5.
Nat Rev Neurosci ; 15(7): 455-65, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24946761

RESUMEN

Malignant gliomas are devastating tumours that frequently kill patients within 1 year of diagnosis. The major obstacle to a cure is diffuse invasion, which enables tumours to escape complete surgical resection and chemo- and radiation therapy. Gliomas use the same tortuous extracellular routes of migration that are travelled by immature neurons and stem cells, frequently using blood vessels as guides. They repurpose ion channels to dynamically adjust their cell volume to accommodate to narrow spaces and breach the blood-brain barrier through disruption of astrocytic endfeet, which envelop blood vessels. The unique biology of glioma invasion provides hitherto unexplored brain-specific therapeutic targets for this devastating disease.


Asunto(s)
Barrera Hematoencefálica/patología , Neoplasias Encefálicas/patología , Glioma/patología , Animales , Neoplasias Encefálicas/genética , Movimiento Celular/fisiología , Glioma/genética , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología
6.
Epilepsia ; 60(7): 1365-1377, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31211419

RESUMEN

OBJECTIVE: Currently prescribed antiepileptic drugs (AEDs) are ineffective in treating approximately 30% of epilepsy patients. Sulfasalazine (SAS) is an US Food and Drug Administration (FDA)-approved drug for the treatment of Crohn disease that has been shown to inhibit the cystine/glutamate antiporter system xc- (SXC) and decrease tumor-associated seizures. This study evaluates the effect of SAS on distinct pharmacologically induced network excitability and determines whether it can further decrease hyperexcitability when administered with currently prescribed AEDs. METHODS: Using in vitro cortical mouse brain slices, whole-cell patch-clamp recordings were made from layer 2/3 pyramidal neurons. Epileptiform activity was induced with bicuculline (bic), 4-aminopyridine (4-AP) and magnesium-free (Mg2+ -free) solution to determine the effect of SAS on epileptiform events. In addition, voltage-sensitive dye (VSD) recordings were performed to characterize the effect of SAS on the spatiotemporal spread of hyperexcitable network activity and compared to currently prescribed AEDs. RESULTS: SAS decreased evoked excitatory postsynaptic currents (eEPSCs) and increased the decay kinetics of evoked inhibitory postsynaptic currents (eIPSCs) in layer 2/3 pyramidal neurons. Although application of SAS to bic and Mg2+ -free-induced epileptiform activity caused a decrease in the duration of epileptiform events, SAS completely blocked 4-AP-induced epileptiform events. In VSD recordings, SAS decreased VSD optical signals induced by 4-AP. Co-application of SAS with the AED topiramate (TPM) caused a significantly further decrease in the spatiotemporal spread of VSD optical signals. SIGNIFICANCE: Taken together this study provides evidence that inhibition of SXC by SAS can decrease network hyperexcitability induced by three distinct pharmacologic agents in the superficial layers of the cortex. Furthermore, SAS provided additional suppression of 4-AP-induced network activity when administered with the currently prescribed AED TPM. These findings may serve as a foundation to assess the potential for SAS or other compounds that selectively target SXC as an adjuvant treatment for epilepsy.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia/tratamiento farmacológico , Sulfasalazina/uso terapéutico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Epilepsia/fisiopatología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Sulfasalazina/farmacología
7.
J Neurosci ; 35(8): 3330-45, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25716834

RESUMEN

Epilepsy is one of the most common chronic neurologic diseases, yet approximately one-third of affected patients do not respond to anticonvulsive drugs that target neurons or neuronal circuits. Reactive astrocytes are commonly found in putative epileptic foci and have been hypothesized to be disease contributors because they lose essential homeostatic capabilities. However, since brain pathology induces astrocytes to become reactive, it is difficult to distinguish whether astrogliosis is a cause or a consequence of epileptogenesis. We now present a mouse model of genetically induced, widespread chronic astrogliosis after conditional deletion of ß1-integrin (Itgß1). In these mice, astrogliosis occurs in the absence of other pathologies and without BBB breach or significant inflammation. Electroencephalography with simultaneous video recording revealed that these mice develop spontaneous seizures during the first six postnatal weeks of life and brain slices show neuronal hyperexcitability. This was not observed in mice with neuronal-targeted ß1-integrin deletion, supporting the hypothesis that astrogliosis is sufficient to induce epileptic seizures. Whole-cell patch-clamp recordings from astrocytes further suggest that the heightened excitability was associated with impaired astrocytic glutamate uptake. Moreover, the relative expression of the cation-chloride cotransporters (CCC) NKCC1 (Slc12a2) and KCC2 (Slc12a5), which are responsible for establishing the neuronal Cl(-) gradient that governs GABAergic inhibition were altered and the NKCC1 inhibitor bumetanide eliminated seizures in a subgroup of mice. These data suggest that a shift in the relative expression of neuronal NKCC1 and KCC2, similar to that observed in immature neurons during development, may contribute to astrogliosis-associated seizures.


Asunto(s)
Astrocitos/metabolismo , Gliosis/metabolismo , Integrina beta1/metabolismo , Convulsiones/metabolismo , Potenciales de Acción , Animales , Astrocitos/fisiología , Barrera Hematoencefálica/metabolismo , Células Cultivadas , Gliosis/fisiopatología , Ácido Glutámico/metabolismo , Integrina beta1/genética , Ratones , Neuronas/metabolismo , Neuronas/fisiología , Convulsiones/etiología , Convulsiones/patología , Convulsiones/fisiopatología , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Simportadores/genética , Simportadores/metabolismo , Cotransportadores de K Cl
8.
Eur Biophys J ; 45(7): 635-648, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27424110

RESUMEN

Malignant gliomas are devastating tumors, frequently killing those diagnosed in little over a year. The profuse infiltration of glioma cells into healthy tissue surrounding the main tumor mass is one of the major obstacles limiting the improvement of patient survival. Migration along the abluminal side of blood vessels is one of the salient features of glioma cell invasion. Invading glioma cells are attracted to the vascular network, in part by the neuropeptide bradykinin, where glioma cells actively modify the gliovascular interface and undergo volumetric alterations to navigate the confined space. Critical to these volume modifications is a proposed hydrodynamic model that involves the flux of ions in and out of the cell, followed by osmotically obligated water. Ion and water channels expressed by the glioma cell are essential in this model of invasion and make opportune therapeutic targets. Lastly, there is growing evidence that vascular-associated glioma cells are able to control the vascular tone, presumably to free up space for invasion and growth. The unique mechanisms that enable perivascular glioma invasion may offer critical targets for therapeutic intervention in this devastating disease. Indeed, a chloride channel-blocking peptide has already been successfully tested in human clinical trials.


Asunto(s)
Vasos Sanguíneos/patología , Glioma/metabolismo , Glioma/patología , Canales Iónicos/metabolismo , Animales , Transporte Biológico , Tamaño de la Célula , Glioma/irrigación sanguínea , Humanos , Invasividad Neoplásica
9.
Brain ; 138(Pt 12): 3716-33, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26598495

RESUMEN

Reduced cerebral blood flow impairs cognitive function and ultimately causes irreparable damage to brain tissue. The gliovascular unit, composed of neural and vascular cells, assures sufficient blood supply to active brain regions. Astrocytes, vascular smooth muscle cells, and pericytes are important players within the gliovascular unit modulating vessel diameters. While the importance of the gliovascular unit and the signals involved in regulating local blood flow to match neuronal activity is now well recognized, surprisingly little is known about this interface in disease. Alzheimer's disease is associated with reduced cerebral blood flow. Here, we studied how the gliovascular unit is affected in a mouse model of Alzheimer's disease, using a combination of ex vivo and in vivo imaging approaches. We specifically labelled vascular amyloid in living mice using the dye methoxy-XO4. We elicited vessel responses ex vivo using either pharmacological stimuli or cell-specific calcium uncaging in vascular smooth muscle cells or astrocytes. Multi-photon in vivo imaging through a cranial window allowed us to complement our ex vivo data in the presence of blood flow after label-free optical activation of vascular smooth muscle cells in the intact brain. We found that vascular amyloid deposits separated astrocyte end-feet from the endothelial vessel wall. High-resolution 3D images demonstrated that vascular amyloid developed in ring-like structures around the vessel circumference, essentially forming a rigid cast. Where vascular amyloid was present, stimulation of astrocytes or vascular smooth muscle cells via ex vivo Ca(2+) uncaging or in vivo optical activation produced only poor vascular responses. Strikingly, vessel segments that were unaffected by vascular amyloid responded to the same extent as vessels from age-matched control animals. We conclude that while astrocytes can still release vasoactive substances, vascular amyloid deposits render blood vessels rigid and reduce the dynamic range of affected vessel segments. These results demonstrate a mechanism that could account in part for the reduction in cerebral blood flow in patients with Alzheimer's disease.media-1vid110.1093/brain/awv327_video_abstractawv327_video_abstract.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Amiloidosis/patología , Amiloidosis/fisiopatología , Astrocitos/patología , Encéfalo/irrigación sanguínea , Encéfalo/fisiopatología , Músculo Liso Vascular/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Astrocitos/ultraestructura , Encéfalo/ultraestructura , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Transgénicos , Músculo Liso Vascular/ultraestructura , Placa Amiloide/patología , Placa Amiloide/fisiopatología
11.
Glia ; 63(1): 23-36, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25066727

RESUMEN

Seizures frequently accompany gliomas and often escalate to peritumoral epilepsy. Previous work revealed the importance of tumor-derived excitatory glutamate (Glu) release mediated by the cystine-glutamate transporter (SXC) in epileptogenesis. We now show a novel contribution of GABAergic disinhibition to disease pathophysiology. In a validated mouse glioma model, we found that peritumoral parvalbumin-positive GABAergic inhibitory interneurons are significantly reduced, corresponding with deficits in spontaneous and evoked inhibitory neurotransmission. Most remaining peritumoral neurons exhibit elevated intracellular Cl(-) concentration ([Cl(-) ]i ) and consequently depolarizing, excitatory gamma-aminobutyric acid (GABA) responses. In these neurons, the plasmalemmal expression of KCC2, which establishes the low [Cl(-) ]i required for GABAA R-mediated inhibition, is significantly decreased. Interestingly, reductions in inhibition are independent of Glu release, but the presence of both decreased inhibition and decreased SXC expression is required for epileptogenesis. We suggest GABAergic disinhibition renders peritumoral neuronal networks hyper-excitable and susceptible to seizures triggered by excitatory stimuli, and propose KCC2 as a therapeutic target.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Epilepsia/metabolismo , Glioma/metabolismo , Receptores de GABA-A/metabolismo , Simportadores/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Neoplasias Encefálicas/complicaciones , Epilepsia/etiología , Femenino , Glioma/complicaciones , Interneuronas/metabolismo , Masculino , Ratones , Neuronas/metabolismo , Transmisión Sináptica/fisiología , Cotransportadores de K Cl
12.
Cereb Cortex ; 24(9): 2388-400, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23585521

RESUMEN

From the subventricular zone (SVZ), neuronal precursor cells (NPCs), called neuroblasts, migrate through the rostral migratory stream (RMS) to become interneurons in the olfactory bulb (OB). Ion channels regulate neuronal migration during development, yet their role in migration through the adult RMS is unknown. To address this question, we utilized Nestin-CreER(T2)/R26R-YFP mice to fluorescently label neuroblasts in the adult. Patch-clamp recordings from neuroblasts reveal K(+) currents that are sensitive to intracellular Ca(2+) levels and blocked by clotrimazole and TRAM-34, inhibitors of intermediate conductance Ca(2+)-activated K(+) (KCa3.1) channels. Immunolabeling and electrophysiology show KCa3.1 expression restricted to neuroblasts in the SVZ and RMS, but absent in OB neurons. Time-lapse confocal microscopy in situ showed inhibiting KCa3.1 prolonged the stationary phase of neuroblasts' saltatory migration, reducing migration speed by over 50%. Both migration and KCa3.1 currents could also be inhibited by blocking Ca(2+) influx via transient receptor potential (TRP) channels, which, together with positive immunostaining for transient receptor potential canonical 1 (TRPC1), suggest that TRP channels are an important Ca(2+) source modulating KCa3.1 activity. Finally, injecting TRAM-34 into Nestin-CreER(T2)/R26R-YFP mice significantly reduced the number of neuroblasts that reached the OB, suggesting an important role for KCa3.1 in vivo. These studies describe a previously unrecognized protein in migration of adult NPCs.


Asunto(s)
Encéfalo/fisiología , Movimiento Celular/fisiología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Células-Madre Neurales/fisiología , Animales , Encéfalo/efectos de los fármacos , Calcio/metabolismo , Movimiento Celular/efectos de los fármacos , Fármacos del Sistema Nervioso Central/farmacología , Clotrimazol/farmacología , Femenino , Inmunohistoquímica , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Masculino , Ratones Transgénicos , Microscopía Confocal , Células-Madre Neurales/efectos de los fármacos , Bulbo Olfatorio/efectos de los fármacos , Bulbo Olfatorio/fisiología , Técnicas de Placa-Clamp , Potasio/metabolismo , Pirazoles/farmacología , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/metabolismo , Técnicas de Cultivo de Tejidos
13.
Cell Mol Life Sci ; 71(10): 1839-54, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24281762

RESUMEN

Malignant gliomas are relentless tumors that offer a dismal clinical prognosis. They develop many biological advantages that allow them to grow and survive in the unique environment of the brain. The glutamate transporters system x c (-) and excitatory amino acid transporters (EAAT) are emerging as key players in the biology and malignancy of these tumors. Gliomas manipulate glutamate transporter expression and function to alter glutamate homeostasis in the brain, which supports their own growth, invasion, and survival. As a consequence, malignant cells are able to quickly destroy and invade surrounding normal brain. Recent findings are painting a larger picture of these transporters in glioma biology, and as such are providing opportunities for clinical intervention for patients. This review will detail the current understanding of glutamate transporters in the biology of malignant gliomas and highlight some of the unique aspects of these tumors that make them so devastating and difficult to treat.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Sistema de Transporte de Aminoácidos X-AG/química , Neoplasias Encefálicas/patología , Glioma/patología , Ácido Glutámico/metabolismo , Glutatión/metabolismo , Humanos , Receptores de Glutamato/química , Receptores de Glutamato/metabolismo
14.
J Neurosci ; 33(4): 1427-40, 2013 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-23345219

RESUMEN

Previous reports demonstrate that cell migration in the nervous system is associated with stereotypic changes in intracellular calcium concentration ([Ca(2+)](i)), yet the target of these changes are essentially unknown. We examined chemotactic migration/invasion of human gliomas to study how [Ca(2+)](i) regulates cellular movement and to identify downstream targets. Gliomas are primary brain cancers that spread exclusively within the brain, frequently migrating along blood vessels to which they are chemotactically attracted by bradykinin. Using simultaneous fura-2 Ca(2+) imaging and amphotericin B perforated patch-clamp electrophysiology, we find that bradykinin raises [Ca(2+)](i) and induces a biphasic voltage response. This voltage response is mediated by the coordinated activation of Ca(2+)-dependent, TRAM-34-sensitive K(Ca)3.1 channels, and Ca(2+)-dependent, 4,4'-diisothiocyanato-stilbene-2,2'-disulfonic acid (DIDS)-sensitive and gluconate-sensitive Cl(-) channels. A significant portion of these Cl(-) currents can be attributed to Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) activation of ClC-3, a voltage-gated Cl(-) channel/transporter, because pharmacological inhibition of CaMKII or shRNA-mediated knockdown of ClC-3 inhibited Ca(2+)-activated Cl(-) currents. Western blots show that K(Ca)3.1 and ClC-3 are expressed in tissue samples obtained from patients diagnosed with grade IV gliomas. Both K(Ca)3.1 and ClC-3 colocalize to the invading processes of glioma cells. Importantly, inhibition of either channel abrogates bradykinin-induced chemotaxis and reduces tumor expansion in mouse brain slices in situ. These channels should be further explored as future targets for anti-invasive drugs. Furthermore, these data elucidate a novel mechanism placing cation and anion channels downstream of ligand-mediated [Ca(2+)](i) increases, which likely play similar roles in other migratory cells in the nervous system.


Asunto(s)
Bradiquinina/metabolismo , Quimiotaxis/fisiología , Canales de Cloruro/metabolismo , Glioma/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Invasividad Neoplásica , Western Blotting , Bradiquinina/farmacología , Línea Celular Tumoral , Quimiotaxis/efectos de los fármacos , Humanos , Inmunohistoquímica , Técnicas de Placa-Clamp
15.
Am J Physiol Cell Physiol ; 306(5): C493-505, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24380845

RESUMEN

Since the seminal studies of Otto Warburg in the 1920s, it has been widely recognized that cancers grow glycolytically, even in the presence of oxygen. This generates an abundance of protons in a gradient across most solid tumors with an acidic core and an alkaline rim. Whether and how this proton gradient may also serve in an autocrine fashion in these tumors is unclear. We demonstrate that human glioma cells form spheroids that act as a viable three-dimensional tumor model, forming physiologically relevant extracellular pH (pHe) and cell proliferation gradients. Using fluorescent cell cycle trackers, we determined that the rate of cell proliferation is directly dependent on pHe and that cells adjust their growth rate according to their position within the pH gradient. We further show that glioma cells sense pH via H(+)-sensitive K(+) channels, which translate changes in pH into changes in membrane voltage. These channels are tonically active and blocked by acidic pHe, quinine, and ruthenium red. Blockade of this K(+) conductance by acidic pHe or drug inhibition depolarized glioma cells and tumor spheroids and prevented their passage through the hyperpolarization-dependent G1-to-S phase cell cycle checkpoint, thereby inhibiting cell division. In this way, pHe directly determines the proliferative state of glioma cells.


Asunto(s)
Comunicación Autocrina , Neoplasias Encefálicas/metabolismo , Proliferación Celular , Glioma/metabolismo , Activación del Canal Iónico , Canales de Potasio/metabolismo , Comunicación Autocrina/efectos de los fármacos , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular , Glioma/patología , Humanos , Concentración de Iones de Hidrógeno , Activación del Canal Iónico/efectos de los fármacos , Cinética , Potenciales de la Membrana , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/efectos de los fármacos , Transducción de Señal , Esferoides Celulares
16.
J Physiol ; 592(22): 5109-27, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25194042

RESUMEN

The molecular and cellular mechanisms governing cell motility and directed migration in response to the neuropeptide bradykinin are largely unknown. Here, we demonstrate that human glioma cells whose migration is guided by bradykinin generate bleb-like protrusions. We found that activation of the B2 receptor leads to a rise in free Ca(2+) from internal stores that activates actomyosin contraction and subsequent cytoplasmic flow into protrusions forming membrane blebs. Furthermore Ca(2+) activates Ca(2+)-dependent K(+) and Cl(-) channels, which participate in bleb regulation. Treatment of gliomas with bradykinin in situ increased glioma growth by increasing the speed of cell migration at the periphery of the tumour mass. To test if bleb formation is related to bradykinin-promoted glioma invasion we blocked glioma migration with blebbistatin, a blocker of myosin kinase II, which is necessary for proper bleb retraction. Our findings suggest a pivotal role of bradykinin during glioma invasion by stimulating amoeboid migration of glioma cells.


Asunto(s)
Bradiquinina/farmacología , Encéfalo/metabolismo , Movimiento Celular , Extensiones de la Superficie Celular/efectos de los fármacos , Glioma/metabolismo , Neuronas/efectos de los fármacos , Animales , Encéfalo/patología , Calcio/metabolismo , Línea Celular Tumoral , Citoesqueleto/metabolismo , Glioma/patología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Invasividad Neoplásica , Neuronas/metabolismo , Neuronas/patología , Canales de Potasio/metabolismo , Canales de Sodio/metabolismo
17.
Glia ; 62(6): 971-81, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24585442

RESUMEN

Glioblastoma multiforme are highly motile primary brain tumors. Diffuse tissue invasion hampers surgical resection leading to poor patient prognosis. Recent studies suggest that intracellular Ca(2+) acts as a master regulator for cell motility and engages a number of downstream signals including Ca(2+) -activated ion channels. Querying the REepository of Molecular BRAin Neoplasia DaTa (REMBRANDT), an annotated patient gene database maintained by the National Cancer Institute, we identified the intermediate conductance Ca(2+) -activated K(+) channels, KCa3.1, being overexpressed in 32% of glioma patients where protein expression significantly correlated with poor patient survival. To mechanistically link KCa3.1 expression to glioma invasion, we selected patient gliomas that, when propagated as xenolines in vivo, present with either high or low KCa3.1 expression. In addition, we generated U251 glioma cells that stably express an inducible knockdown shRNA to experimentally eliminate KCa3.1 expression. Subjecting these cells to a combination of in vitro and in situ invasion assays, we demonstrate that KCa3.1 expression significantly enhances glioma invasion and that either specific pharmacological inhibition with TRAM-34 or elimination of the channel impairs invasion. Importantly, after intracranial implantation into SCID mice, ablation of KCa3.1 with inducible shRNA resulted in a significant reduction in tumor invasion into surrounding brain in vivo. These results show that KCa3.1 confers an invasive phenotype that significantly worsens a patient's outlook, and suggests that KCa3.1 represents a viable therapeutic target to reduce glioma invasion.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Movimiento Celular/fisiología , Glioma/metabolismo , Glioma/patología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/fisiología , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Bases de Datos Genéticas , Femenino , Glioma/genética , Humanos , Masculino , Ratones , Ratones SCID , Invasividad Neoplásica/patología
18.
J Neurosci Methods ; 407: 110144, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38670535

RESUMEN

BACKGROUND: The enteric nervous system (ENS) is comprised of neurons, glia, and neural progenitor cells that regulate essential gastrointestinal functions. Advances in high-efficiency enteric neuron culture would facilitate discoveries surrounding ENS regulatory processes, pathophysiology, and therapeutics. NEW METHOD: Development of a simple, robust, one-step method to culture murine enteric neurospheres in a 3D matrix that supports neural growth and differentiation. RESULTS: Myenteric plexus cells isolated from the entire length of adult murine small intestine formed ≥3000 neurospheres within 7 days. Matrigel-embedded neurospheres exhibited abundant neural stem and progenitor cells expressing Sox2, Sox10 and Msi1 by day 4. By day 5, neural progenitor cell marker Nestin appeared in the periphery of neurospheres prior to differentiation. Neurospheres produced extensive neurons and neurites, confirmed by Tubulin beta III, PGP9.5, HuD/C, and NeuN immunofluorescence, including neural subtypes Calretinin, ChAT, and nNOS following 8 days of differentiation. Individual neurons within and external to neurospheres generated depolarization induced action potentials which were inhibited in the presence of sodium channel blocker, Tetrodotoxin. Differentiated neurospheres also contained a limited number of glia and endothelial cells. COMPARISON WITH EXISTING METHODS: This novel one-step neurosphere growth and differentiation culture system, in 3D format (in the presence of GDNF, EGF, and FGF2), allows for ∼2-fold increase in neurosphere count in the derivation of enteric neurons with measurable action potentials. CONCLUSION: Our method describes a novel, robust 3D culture of electrophysiologically active enteric neurons from adult myenteric neural stem and progenitor cells.


Asunto(s)
Plexo Mientérico , Neuronas , Animales , Plexo Mientérico/citología , Plexo Mientérico/fisiología , Neuronas/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Célula/métodos , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Células-Madre Neurales/efectos de los fármacos , Diferenciación Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Células Cultivadas , Potenciales de Acción/fisiología , Potenciales de Acción/efectos de los fármacos , Laminina/farmacología , Combinación de Medicamentos , Proteoglicanos/farmacología , Masculino , Neurogénesis/fisiología , Neurogénesis/efectos de los fármacos , Colágeno
19.
Nat Neurosci ; 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39020018

RESUMEN

Perineuronal nets (PNNs) are densely packed extracellular matrices that cover the cell body of fast-spiking inhibitory neurons. PNNs stabilize synapses inhibiting synaptic plasticity. Here we show that synaptic terminals of fast-spiking interneurons localize to holes in the PNNs in the adult mouse somatosensory cortex. Approximately 95% of holes in the PNNs contain synapses and astrocytic processes expressing Kir4.1, glutamate and GABA transporters. Hence, holes in the PNNs contain tripartite synapses. In the adult mouse brain, PNN degradation causes an expanded astrocytic coverage of the neuronal somata without altering the axon terminals. The loss of PNNs impairs astrocytic transmitter and potassium uptake, resulting in the spillage of glutamate into the extrasynaptic space. Our data show that PNNs and astrocytes cooperate to contain synaptically released signals in physiological conditions. Their combined action is altered in mouse models of Alzheimer's disease and epilepsy where PNNs are disrupted.

20.
Neuroscientist ; 29(2): 158-165, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-33754906

RESUMEN

Once strictly the domain of medical and graduate education, neuroscience has made its way into the undergraduate curriculum with over 230 colleges and universities now offering a bachelor's degree in neuroscience. The disciplinary focus on the brain teaches students to apply science to the understanding of human behavior, human interactions, sensation, emotions, and decision making. In this article, we encourage new and existing undergraduate neuroscience programs to envision neuroscience as a broad discipline with the potential to develop competencies suitable for a variety of careers that reach well beyond research and medicine. This article describes our philosophy and illustrates a broad-based undergraduate degree in neuroscience implemented at a major state university, Virginia Tech. We highlight the fact that the research-centered Experimental Neuroscience major is least popular of our four distinct majors, which underscores our philosophy that undergraduate neuroscience can cater to a different audience than traditionally thought.


Asunto(s)
Neurociencias , Humanos , Neurociencias/educación , Curriculum , Estudiantes , Universidades , Encéfalo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA